• No results found

VU Research Portal

N/A
N/A
Protected

Academic year: 2021

Share "VU Research Portal"

Copied!
21
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

VU Research Portal

Acquired Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors

in Non-Small Cell Lung Cancer

Kuiper, J.L.

2016

document version

Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)

Kuiper, J. L. (2016). Acquired Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in

Non-Small Cell Lung Cancer.

General rights

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain

• You may freely distribute the URL identifying the publication in the public portal ? Take down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address:

(2)

Introduction

(3)
(4)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

1

LUNG CANCER

Incidence and risk factors

Lung cancer is the leading cause of cancer-related deaths worldwide (1). In 2015, in the

Netherlands, more than 12.000 patients were diagnosed with lung cancer and almost 11.000

patients died of the disease (2). The lifetime risk of being diagnosed with cancer of the lung

is approximately 6% (3). The most important risk factor for lung cancer is smoking; 85% of

all newly diagnosed cases of lung cancer is associated with smoking (4). Yet, also in

never-smokers, lung cancer ranks the seventh cause of cancer-related mortality worldwide (5).

Geographical variations, exposure to occupational and domestic carcinogens, hormonal and

environmental factors, as well as genetic predisposition may play a role as etiologic risk factors

(6). The relatively high risk of developing lung cancer is probably also caused by stochastic

effects associated with the lifetime number of stem cell divisions of lung tissue (7).

Classification of lung cancer

Lung cancer is histologically classified according to guidelines of the World Health Organization

(WHO) (8) (Figure 1A). The predominant histological subtype of lung cancer is non-small cell

lung cancer (NSCLC); approximately 80% of lung cancer patients is diagnosed with this type

of lung cancer (9). The remaining part concerns small-cell lung cancer (SCLC) (14-18%) and

other, more rare histological subtypes (i.e., mesothelioma and neuro-endocrine tumours)

(6%) (2, 10). Among NSCLC, the predominating histological subtype is adenocarcinoma

(approximately 40 – 45%) (11). Squamous cell carcinoma is diagnosed in 25 – 31% of

NSCLC-patients and 9 – 18% of NSCLC is of the large-cell carcinoma subtype (10, 12).

(5)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

R35

R36

R37

R38

R39

Chapter 1 Fig. 1A Fig. 1B

Lung cancer

NSCLC

SCLC

Adenocarcinoma Squamous cell carcinoma Large cell carcinoma

Classification based on histology Classification of lung adeno-carcinoma based on molecular genetics

Figure 1: Classification of lung cancer

Fig. 1A: Classification of lung cancer based on histology.

Fig. 1B: Classification of lung adenocarcinoma based on molecular features. The relative frequency of major driver mutations in signaling molecules in lung adenocarcinoma is shown.

NSCLC: non-small cell lung cancer. SCLC: small cell lung cancer. EGFR: epidermal growth factor receptor mutation. KRAS: Kirsten rat sarcoma. ALK: echinoderm-associated protein-like 4 (EML4) anaplastic lymphoma kinase (ALK) translocation. ROS: ROS1 rearrangement. RET: RET fusion gene. HER2: HER2 mutation. BRAF: BRAF mutation.

(6)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

1

Genetics in lung cancer

In lung cancer, the most prevalent mutations are found in the gene encoding the Kirsten

rat sarcoma viral oncogene homolog (KRAS)- (29%), the epidermal growth factor receptor

(EGFR)- (11%), V-Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF)- (2%) and human

epidermal growth factor receptor 2 (HER2)- gene (1%) (16). Rearrangements occur in the

anaplastic lymphoma kinase (ALK) gene (5%) (16), Ret proto-oncogene (RET) gene (1%) (17)

and C-Ros Oncogene 1 (ROS1) (1%) (18). Amplification has been described in HER2 (18%) (1,

19) and Hepatocyte Growth Factor Receptor MET (2-4%) (20). Nevertheless, there is a wide

variance in incidence of these mutations between geographical regions worldwide. These

oncogenic drivers occur predominantly in adenocarcinoma and existence is usually mutually

exclusive.

Epidermal growth factor receptor

The protein EGFR is a member of the HER family, one of the 20 families of transmembrane

receptor tyrosine kinases. Tyrosine kinases are enzymes that can attach phosphate groups to

other amino acids. Phosphorylation of proteins by tyrosine kinases is an important mechanism

for intracellular communication (signal transduction) and regulating cellular activity, such as

cell division.

(7)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

R35

R36

R37

R38

R39

Chapter 1     N-­‐lobe  C-­‐lobe

ATP  binding  cleft

Extracellular Intracellular Transmembrane TK-­‐domain Exon  18 Exon  19 Exon  21 Exon  20 Exon  22 Exon  23 Exon  24 Region  where   relevant  EGFR-­‐ mutations  are   located

EGF-­‐receptor

EGFR  gene

Figure legend:

The EGF receptor.

Figure 2

Figure 2: The EGF Receptor

EGFR-mutations in cancer

Excessive signalling through the members of the HER-receptor family is associated with

several types of cancer. Mutations in EGFR have been reported in a variety of cancers,

including anal cancer, glioblastoma multiforme and lung cancer (23, 24). In lung cancer, 90%

of all EGFR-mutations are represented by mutations in two regions, often referred to as

the ‘classic’ activating EGFR-mutations (25). These mutations concern in-frame deletions in

exon 19 (usually around the amino acids Glycine-Leucine-Arginine-Glycine-Alanine (ELREA),

residues 746-750) (45 – 50%) and the Leu858Arg (L858R) substitution, resulting from a point

mutation in exon 21 (40 – 45%) (26). Classic EGFR-mutations increase the kinase activity

of the protein, thereby continuously activating the downstream pro-survival pathways in

absence of ligand-binding (27). The remaining 10% of EGFR-mutations concern so-called

‘non-classic mutations’ (or: ‘uncommon’ mutations) in exons 18 – 21.

EGFR-mutations are reported to occur almost exclusively in non-squamous NSCLC (8). 9.4%

of Caucasian NSCLC-patients and up to 47.9% of Asian NSCLC-patients carry EGFR-mutations

in their tumours (16, 28). Regardless of ethnicity and histology, clinical characteristics that are

associated with EGFR-mutations are non-smoking (14 – 56%) and female gender (20 – 62%)

(26, 29-32), however EGFR-mutations are also reported in males (1 – 19%) and smokers (3 –

14%) as well (26).

Stage, treatment options, survival and prognosis of NSCLC

(8)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

1

Patients with local (stage IA – IIB) and locally advanced NSCLC (IIIA) can be curatively

treated with surgery, (stereotactic) radiotherapy and/or chemotherapy. However, due

to a lack of symptoms in the early stages of disease, the vast majority of NSCLC-patients

(more than 80%) have advanced-stage disease (stage IIIB – IV) at time of first diagnosis. For

advanced-stage NSCLC patients there are currently no curative treatment options available

(4). Nonetheless, (combinations of) new treatment modalities have been investigated in

recent years and have improved treatment outcomes of subsets of advanced-stage NSCLC.

Prognosis of advanced-stage lung cancer is to some extent dependent on biological and

clinical factors, such as weight loss, histological and molecular subtype, and performance

status (PS) (34). Despite improvement in the treatment of advanced-stage NSCLC, overall

survival (OS) is still poor. Median OS after diagnosis of stage IV NSCLC adenocarcinoma is

12.6 months (35) and the five-year survival rate of unselected patients with stage IV NSCLC

is less than 5% (1). EGFR-mutated NSCLC-patients have a better prognosis compared to the

unselected NSCLC-population; for these patients a five-year survival rate of 14% has been

reported (36).

  Figure 3

Figure 3: EGFR signalling pathways.

(9)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

R35

R36

R37

R38

R39

Chapter 1

Treatment of advanced stage lung cancer

Since the discovery of platinum-based chemotherapeutic agents as anti-cancer treatment

in the 1970s, these drugs have been the standard first-line treatment for advanced-stage

lung cancer (37). Until the 1990s all advanced-stage lung cancer patients were treated with a

platinum-based chemotherapeutic regimen (usually combined with etoposide). From 2000,

the distinction between SCLC and NSCLC became clinically relevant, when it was demonstrated

that specific chemotherapeutic regimens have different efficacy in SCLC and NSCLC (38).

First-line treatment with chemotherapy improves overall survival of stage IV

NSCLC-patients by 9% (39). The optimal duration of first-line platinum-based chemotherapy

is considered to be four cycles (39, 40). A two-agent regimen of cytotoxic chemotherapy

provides the optimal effect considering tumour response and overall survival compared to

one- or three-agent regimens (41). The histological subtype of NSCLC may guide the selection

of the second chemotherapeutic agent, next to the platinum-based chemotherapeutic agent.

Patients with squamous cell carcinoma should not be treated with the third-generation

chemotherapeutic agent pemetrexed, but receive a different agent (e.g. gemcitabine) (42).

(10)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

1

TARGETED TREATMENT OF EGFR-MUTATED NSCLC-PATIENTS

EGFR-inhibition in lung cancer

EGFR-tyrosine kinase inhibitors (TKIs) are small molecule TKIs that bind to the tyrosine kinase

domain of the EGFR. The catalytic activity of the tyrosine kinase is blocked by the competition

of these agents with adenosine triphosphate (ATP). Gefitinib (ZD1839, Iressa) and erlotinib

(OSI-774, Tarceva) bind reversibly to the EGFR and were the first EGFR-TKIs to be registered

for lung cancer treatment. The EGFR-TKI afatinib (BIBW2992, Giotrif) is a pan-HER binding

agent and was the subsequent EGFR-TKI to be registered for the treatment of lung cancer.

In 2003, the Food and Drug Administration (FDA) granted accelerated proof for gefitinib

after evaluation in previously treated NSCLC-patients in two randomized phase II trials (43,

44). Performance status (PS), histology and female gender were clinical parameters that

were associated with a response. Most frequently reported toxicities were acne-like rash

and gastro-intestinal side effects. The second TKI that received FDA-approval for treatment

of patients with advanced stage NSCLC was erlotinib, after evaluation in the randomized,

placebo-controlled, double-blind BR.21 trial (45). Retrospective subset analyses showed that

also adenocarcinoma and smoking status were associated with response to erlotinib.

A phase III trial evaluated gefitinib in previously treated NSCLC-patients (46), but no

difference in survival was detected. However, in this trial, it was suggested that

EGFR-mutations might be the predictive biomarker for response to EGFR-TKIs (47). The INTEREST

trial was the first phase III trial that demonstrated that NSCLC-patients with EGFR-mutations

had longer PFS and higher ORR when treated with gefitinib compared to docetaxel (48).

The discovery that EGFR-mutations were the biomarker for prediction of response to

EGFR-TKI treatment in 2004 (29, 30), led to the initiation of a variety of phase III randomized

trials evaluating EGFR-TKIs in EGFR-mutated NSCLC patients (Table 1). All of these trials

demonstrated the beneficial effect of EGFR-TKIs as compared to cytotoxic chemotherapy in

NSCLC-patients carrying an EGFR-mutation, in terms of prolongation of PFS and improved

response rate. Since then, first-generation EGFR-TKIs are incorporated as first-line treatment

for EGFR-mutated NSCLC-patients in international guidelines (49-51). Erlotinib is also

registered for second-line treatment of unselected NSCLC-patients, based on the results of

the BR.21 trial (45). Despite the evident effect of erlotinib and gefitinib on PFS and response

rates, a beneficial effect on overall survival has never been demonstrated which is probably

due to cross-over of patients to EGFR-TKIs after treatment with chemotherapy (52).

(11)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

R35

R36

R37

R38

R39

Chapter 1

First-line afatinib improved OS in two phase III trials for patients with a exon 19 deletion, but

not for patients with an L858R point mutation (62). However, the results of afatinib in

NSCLC-patients with acquired resistance to erlotinib or gefitinib were disappointing; response rate

was only 8.2% (63) and OS was not prolonged in a placebo-controlled study (64).

Resistance mechanisms in EGFR-TKI treatment

(12)
(13)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

R35

R36

R37

R38

R39

Chapter 1

OUTLINE OF THIS THESIS

Lung cancer is the leading cause of cancer related mortality in the Netherlands, indicating that

novel therapies and improved treatment strategies are urgently needed. Classically, treatment

decisions have been empiric and mainly based upon histology of the tumour. Over the past

decade, it has become evident that subsets of NSCLC can be further defined at the molecular

level by recurrent ‘driver’ mutations. One of such altered genes is EGFR, which is mutated

in approximately 10% of NSCLC-patients in the Netherlands (76). Importantly, targeted

small molecule inhibitors are currently available for EGFR-mutated lung cancer patients and

have become the standard of treatment for these patients. The introduction of these

first-generation EGFR-TKIs in the treatment of lung cancer has been a major step forward, yet

new challenges and questions have arisen as well. This thesis describes the results of clinical

studies conducted among EGFR-mutated NSCLC-patients and NSCLC-patients with acquired

resistance to first-generation EGFR-TKIs. The first part focuses on diagnostics and predictive

markers, and the second part focuses on treatment.

Diagnostics & response prediction

Prevalence of uncommon EGFR-mutations

Studies that report on non-classic EGFR-mutations are limited and data on EGFR-TKI

sensitivity of these mutations is scarce, especially in non-Asian populations. In Chapter 2, we

describe the prevalence and type distribution of non-classic EGFR-mutations among Dutch

EGFR-mutated NSCLC-patients, as well as clinical characteristics and outcomes on EGFR-TKI

treatment in this cohort.

T790M mutation as mechanism of EGFR-TKI-resistance

The T790M mutation is the most frequently detected mechanism of resistance in

EGFR-mutated NSCLC-patients after having acquired resistance to EGFR-TKI treatment. The

mutation is rarely found independently of EGFR-TKI treatment, however some patients in

whom the T790M mutation was detected prior to EGFR-TKI treatment have been reported

(77). Heterogeneity of T790M detection has been described in individual cases, both in time

(78) and between different tumour lesions (79). In Chapter 3, we describe the occurrence

of T790M mutations in a cohort of EGFR-mutated NSCLC patients who were rebiopsied after

having acquired EGFR-TKI-resistance.

Histological transformation and tumour heterogeneity

(14)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

1

has gained interest given its likely role in the development of resistance to treatment. In

Chapter 4a, we describe an EGFR-mutated NSCLC-patient in whom an innovative PET imaging

method was used that could possibly play a role in detecting tumour heterogeneity in the

future.

Histological transformation after treatment with EGFR-TKIs, e.g. transformation from

NSCLC to SCLC, has been described as resistance mechanism, but is infrequently reported

(67). Histological transformation may develop as a result of tumour heterogeneity that was

present prior to treatment initiation. In Chapter 4b we describe a patient who was rebiopsied

after acquired resistance to EGFR-TKI treatment and in whom a different form of histologic

transformation after EGFR-TKI treatment was reported; transition of an adenocarcinoma to

squamous cell carcinoma phenotype.

Serum-based proteomic biomarkers

Although the presence of an EGFR-mutation in the tumour is predictive for response on

EGFR-TKI treatment, some EGFR-wild type (WT) patients experience prolongation of PFS

when treated with EGFR-TKI treatment (45). Moreover, in some patients assessment of

tumour mutation-status is either not possible or it is not possible to obtain (new)

tumour-tissue. Therefore, it would be helpful to have a less-invasive biomarker test available that is

predictive for response to EGFR-TKI treatment.

VeriStrat is a serum-based proteomic test that is commercially available in the United

States and has shown to predict outcome after treatment with EGFR-TKIs (81-85). Chapter 5

describes the results of the VeriStrat-test in a cohort of unselected NSCLC-patients who were

treated with the combination of erlotinib and sorafenib.

Treatment

Present-day challenges in EGFR-TKI-resistant NSCLC

Since two decennia, response to anti-cancer treatment is measured according to the response

evaluation criteria in solid tumors (RECIST) (86). One of the important issues is whether

traditional response evaluation criteria are still applicable in the treatment with targeted

agents. This issue is reviewed in Chapter 6, as well as the mechanisms of resistance and

different treatment strategies in EGFR-mutated NSCLC with acquired EGFR-TKI resistance.

Leptomeningeal metastases

(15)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

R35

R36

R37

R38

R39

Chapter 1

the compartment of the cerebrospinal fluid (89). It is considered to be a complication with

poor prognosis and rapid deterioration of performance status (90). In Chapter 7, we describe

diagnosis, treatment and survival of EGFR-mutated NSCLC-patients with leptomeningeal

metastases.

Pulsatile EGFR-TKI treatment

It is believed that, due to the blood-brain barrier (BBB), EGFR-TKIs do not reach therapeutic

concentrations in the intra-central nervous system (CNS) compartment when administered

in standard dose (91). It is hypothesized that with high-dose weekly erlotinib, therapeutic

concentrations in the intra-CNS fluid can be reached (92). In Chapter 8a, we describe

two EGFR-mutated NSCLC-patients who developed leptomeningeal metastases and both

responded to high-dose EGFR-TKI treatment. Interestingly, one patient also had a response

of the intrathoracic lesions that had been resistant for previous chemotherapy.

Since EGFR-TKIs are competitive inhibitors of EGF signalling, it can be hypothesized that

higher doses of the drug might restore the sensitivity. The results of a phase II trial that

evaluated high-dose weekly EGFR-TKI treatment in EGFR-mutated NSCLC-patients who

acquired resistance to standard-dose EGFR-TKI treatment, are described in Chapter 8b.

Afatinib

(16)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

1

REFERENCE LIST

(1) Ferlay J, Soerjomataram I, Ervik M, et al. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality worldwide: IARC CancerBase No.11. Lyan, France: International Agency for Research on Cancer; 2013. Available from: http://globocan.iarc.fr, accessed on 15/05/2016.

(2) IKNL. Cijfers over kanker. Available from: www.cijfersoverkanker.nl/selecties/Dataset_3/ img57385c601df0e, accessed on 15/05/2016.

(3) SEER Cancer Statistics. Available from: http://seer.cancer.gov, accessed on 16/05/2016.

(4) Landelijke werkgroep longtumoren. Niet kleincellig longcarcinoom, landelijke richtlijn. Oncoline. Available from: www.oncoline.nl/niet-kleincellig-longcarcinoom, accessed on 15/05/2016. (5) Couraud S, Zalcman G, Milleron B, Morin F, Souquet PJ. Lung cancer in never smokers--a review.

Eur J Cancer 2012 Jun,48(9), 1299-1311.

(6) Sun S, Schiller JH, Gazdar AF. Lung cancer in never smokers--a different disease. Nat Rev Cancer 2007 Oct,7(10), 778-790.

(7) Tomasetti C, Vogelstein B. Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 2015 Jan 2,347(6217), 78-81.

(8) Travis WD, Brambilla E, Noguchi M, et al. International association for the study of lung cancer/american thoracic society/european respiratory society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol 2011 Feb,6(2), 244-285.

(9) Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin 2011 Mar,61(2), 69-90.

(10) Wahbah M, Boroumand N, Castro C, El-Zeky F, Eltorky M. Changing trends in the distribution of the histologic types of lung cancer: a review of 4,439 cases. Ann Diagn Pathol 2007 Apr,11(2), 89-96.

(11) Devesa SS, Bray F, Vizcaino AP, Parkin DM. International lung cancer trends by histologic type: male:female differences diminishing and adenocarcinoma rates rising. Int J Cancer 2005 Nov 1,117(2), 294-299.

(12) Thunnissen E, van der Oord K, den Bakker M. Prognostic and predictive biomarkers in lung cancer. A review. Virchows Arch 2014 Mar,464(3), 347-358.

(13) Pao W, Hutchinson KE. Chipping away at the lung cancer genome. Nat Med 2012 Mar,18(3), 349-351.

(14) Weinstein IB. Cancer. Addiction to oncogenes--the Achilles heal of cancer. Science 2002 Jul 5,297(5578), 63-64.

(15) Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med 2004 Aug,10(8), 789-799.

(16) Barlesi F, Mazieres J, Merlio JP, et al. Routine molecular profiling of patients with advanced non-small-cell lung cancer: results of a 1-year nationwide programme of the French Cooperative Thoracic Intergroup (IFCT). Lancet 2016 Jan 14.

(17) Takeuchi K, Soda M, Togashi Y, et al. RET, ROS1 and ALK fusions in lung cancer. Nat Med 2012 Mar,18(3), 378-381.

(17)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

R35

R36

R37

R38

R39

Chapter 1

(21) Linggi B, Carpenter G. ErbB receptors: new insights on mechanisms and biology. Trends Cell Biol 2006 Dec,16(12), 649-656.

(22) Oda K, Matsuoka Y, Funahashi A, Kitano H. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol Syst Biol 2005,1, 2005.

(23) Walker F, Abramowitz L, Benabderrahmane D, et al. Growth factor receptor expression in anal squamous lesions: modifications associated with oncogenic human papillomavirus and human immunodeficiency virus. Hum Pathol 2009 Nov,40(11), 1517-1527.

(24) Brennan CW, Verhaak RG, McKenna A, et al. The somatic genomic landscape of glioblastoma. Cell 2013 Oct 10,155(2), 462-477.

(25) Riely GJ, Politi KA, Miller VA, Pao W. Update on epidermal growth factor receptor mutations in non-small cell lung cancer. Clin Cancer Res 2006 Dec 15,12(24), 7232-7241.

(26) Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst 2005 Mar 2,97(5), 339-346.

(27) Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science 2004 Aug 20,305(5687), 1163-1167.

(28) Dearden S, Stevens J, Wu YL, Blowers D. Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap). Ann Oncol 2013 Sep,24(9), 2371-2376.

(29) Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004 May 20,350(21), 2129-2139.

(30) Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004 Jun 4,304(5676), 1497-1500.

(31) Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl

Acad Sci U S A 2004 Sep 7,101(36), 13306-13311.

(32) D’Angelo SP, Pietanza MC, Johnson ML, et al. Incidence of EGFR exon 19 deletions and L858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J Clin Oncol 2011 May 20,29(15), 2066-2070.

(33) Goldstraw P, Crowley J, Chansky K, et al. The IASLC Lung Cancer Staging Project: proposals for the revision of the TNM stage groupings in the forthcoming (seventh) edition of the TNM Classification of malignant tumours. J Thorac Oncol 2007 Aug,2(8), 706-714.

(34) Hoang T, Dahlberg SE, Sandler AB, Brahmer JR, Schiller JH, Johnson DH. Prognostic models to predict survival in non-small-cell lung cancer patients treated with first-line paclitaxel and carboplatin with or without bevacizumab. J Thorac Oncol 2012 Sep,7(9), 1361-1368.

(35) Scagliotti GV, Parikh P, von PJ, et al. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol 2008 Jul 20,26(21), 3543-3551.

(36) Lin JJ, Cardarella S, Lydon CA, et al. Five-Year Survival in EGFR-Mutant Metastatic Lung Adenocarcinoma Treated with EGFR-TKIs. J Thorac Oncol 2016 Apr,11(4), 556-565.

(37) Kelland L. The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer 2007 Aug,7(8), 573-584.

(38) Reck M, Heigener DF, Mok T, Soria JC, Rabe KF. Management of non-small-cell lung cancer: recent developments. Lancet 2013 Aug 24,382(9893), 709-719.

(18)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

1

(40) Park JO, Kim SW, Ahn JS, et al. Phase III trial of two versus four additional cycles in patients who are nonprogressive after two cycles of platinum-based chemotherapy in non small-cell lung cancer. J Clin Oncol 2007 Nov 20,25(33), 5233-5239.

(41) Delbaldo C, Michiels S, Rolland E, et al. Second or third additional chemotherapy drug for non-small cell lung cancer in patients with advanced disease. Cochrane Database Syst Rev 2007,(4), CD004569.

(42) Scagliotti G, Hanna N, Fossella F, et al. The differential efficacy of pemetrexed according to NSCLC histology: a review of two Phase III studies. Oncologist 2009 Mar,14(3), 253-263.

(43) Fukuoka M, Yano S, Giaccone G, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer (The IDEAL 1 Trial) [corrected]. J Clin Oncol 2003 Jun 15,21(12), 2237-2246.

(44) Kris MG, Natale RB, Herbst RS, et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. JAMA 2003 Oct 22,290(16), 2149-2158.

(45) Shepherd FA, Rodrigues PJ, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 2005 Jul 14,353(2), 123-132.

(46) Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer). Lancet 2005 Oct 29,366(9496), 1527-1537.

(47) Hirsch FR, Varella-Garcia M, Bunn PA, Jr., et al. Molecular predictors of outcome with gefitinib in a phase III placebo-controlled study in advanced non-small-cell lung cancer. J Clin Oncol 2006 Nov 1,24(31), 5034-5042.

(48) Kim ES, Hirsh V, Mok T, et al. Gefitinib versus docetaxel in previously treated non-small-cell lung cancer (INTEREST): a randomised phase III trial. Lancet 2008 Nov 22,372(9652), 1809-1818. (49) Keedy VL, Temin S, Somerfield MR, et al. American Society of Clinical Oncology provisional clinical

opinion: epidermal growth factor receptor (EGFR) Mutation testing for patients with advanced non-small-cell lung cancer considering first-line EGFR tyrosine kinase inhibitor therapy. J Clin

Oncol 2011 May 20,29(15), 2121-2127.

(50) National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Non-small cell lung cancer. V4.2016. Available from: http://www.nccn.org/professionals/physician_ gls/f_guidelines.asp#nsclc, accessed on 15/15/2016.

(51) Reck M, Popat S, Reinmuth N, De RD, Kerr KM, Peters S. Metastatic non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2014 Sep,25 Suppl 3, iii27-iii39.

(52) Liang W, Wu X, Fang W, et al. Network meta-analysis of erlotinib, gefitinib, afatinib and icotinib in patients with advanced non-small-cell lung cancer harboring EGFR mutations. PLoS One 2014,9(2), e85245.

(53) Han JY, Park K, Kim SW, et al. First-SIGNAL: first-line single-agent iressa versus gemcitabine and cisplatin trial in never-smokers with adenocarcinoma of the lung. J Clin Oncol 2012 Apr 1,30(10), 1122-1128.

(54) Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 2010 Jun 24,362(25), 2380-2388.

(19)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

R35

R36

R37

R38

R39

Chapter 1

(57) Sequist LV, Yang JC, Yamamoto N, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 2013 Sep 20,31(27), 3327-3334.

(58) Zhou C, Wu YL, Chen G, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 2011 Aug,12(8), 735-742. (59) Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker analyses and final overall survival results

from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J Clin Oncol 2011 Jul 20,29(21), 2866-2874.

(60) Wu YL, Zhou C, Hu CP, et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol 2014 Feb,15(2), 213-222.

(61) Li D, Ambrogio L, Shimamura T, et al. BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models. Oncogene 2008 Aug 7,27(34), 4702-4711.

(62) Yang JC, Wu YL, Schuler M, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 2015 Feb,16(2), 141-51.

(63) Katakami N, Atagi S, Goto K, et al. LUX-Lung 4: a phase II trial of afatinib in patients with advanced non-small-cell lung cancer who progressed during prior treatment with erlotinib, gefitinib, or both. J Clin Oncol 2013 Sep 20,31(27), 3335-3341.

(64) Miller VA, Hirsh V, Cadranel J, et al. Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol 2012 May,13(5), 528-38.

(65) Chong CR, Janne PA. The quest to overcome resistance to EGFR-targeted therapies in cancer. Nat

Med 2013 Nov,19(11), 1389-1400.

(66) Sequist LV, Waltman BA, as-Santagata D, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 2011 Mar 23,3(75), 75ra26. (67) Yu HA, Arcila ME, Rekhtman N, et al. Analysis of Tumor Specimens at the Time of Acquired

Resistance to EGFR-TKI Therapy in 155 Patients with EGFR-Mutant Lung Cancers. Clin Cancer Res 2013 Apr 15,19(8):2240-7.

(68) Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med 2005 Feb 24,352(8), 786-792.

(69) Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2005 Mar,2(3), e73.

(70) Yun CH, Mengwasser KE, Toms AV, et al. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci U S A 2008 Feb 12,105(6), 2070-2075.

(71) Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007 May 18,316(5827), 1039-1043.

(72) Takezawa K, Pirazzoli V, Arcila ME, et al. HER2 amplification: a potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov 2012 Oct,2(10), 922-933.

(73) Yonesaka K, Zejnullahu K, Okamoto I, et al. Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab. Sci Transl Med 2011 Sep 7,3(99), 99ra86. (74) Mitsudomi T, Morita S, Yatabe Y, et al. Gefitinib versus cisplatin plus docetaxel in patients with

(20)

R1

R2

R3

R4

R5

R6

R7

R8

R9

R10

R11

R12

R13

R14

R15

R16

R17

R18

R19

R20

R21

R22

R23

R24

R25

R26

R27

R28

R29

R30

R31

R32

R33

R34

1

(75) Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009 Sep 3,361(10), 947-957.

(76) Smits AJ, Kummer JA, Hinrichs JW, et al. EGFR and KRAS mutations in lung carcinomas in the Dutch population: increased EGFR mutation frequency in malignant pleural effusion of lung adenocarcinoma. Cell Oncol (Dordr) 2012 Jun,35(3), 189-196.

(77) Yu HA, Arcila ME, Hellmann MD, Kris MG, Ladanyi M, Riely GJ. Poor response to erlotinib in patients with tumors containing baseline EGFR T790M mutations found by routine clinical molecular testing. Ann Oncol 2014 Feb,25(2), 423-428.

(78) Hata A, Katakami N, Kaji R, Fujita S, Imai Y. Does T790M disappear? Successful gefitinib rechallenge after T790M disappearance in a patient with EGFR-mutant non-small-cell lung cancer. J Thorac

Oncol 2013 Mar,8(3), e27-e29.

(79) Hata A, Masago K, Katakami N, Imai Y, Yatabe Y. Spatiotemporal T790M Heterogeneity in a Patient with EGFR-Mutant Non-Small-Cell Lung Cancer. J Thorac Oncol 2014 Aug,9(8), e64-e65.

(80) Fisher R, Pusztai L, Swanton C. Cancer heterogeneity: implications for targeted therapeutics. Br J

Cancer 2013 Feb 19,108(3), 479-485.

(81) Amann JM, Lee JW, Roder H, et al. Genetic and proteomic features associated with survival after treatment with erlotinib in first-line therapy of non-small cell lung cancer in Eastern Cooperative Oncology Group 3503. J Thorac Oncol 2010 Feb,5(2), 169-178.

(82) Carbone DP, Ding K, Roder H, et al. Prognostic and predictive role of the VeriStrat plasma test in patients with advanced non-small-cell lung cancer treated with erlotinib or placebo in the NCIC Clinical Trials Group BR.21 trial. J Thorac Oncol 2012 Nov,7(11), 1653-1660.

(83) Chung CH, Seeley EH, Roder H, et al. Detection of tumor epidermal growth factor receptor pathway dependence by serum mass spectrometry in cancer patients. Cancer Epidemiol Biomarkers Prev 2010 Feb,19(2), 358-365.

(84) Stinchcombe TE, Roder J, Peterman AH, et al. A retrospective analysis of VeriStrat status on outcome of a randomized phase II trial of first-line therapy with gemcitabine, erlotinib, or the combination in elderly patients (age 70 years or older) with stage IIIB/IV non-small-cell lung cancer. J Thorac Oncol 2013 Apr,8(4), 443-451.

(85) Taguchi F, Solomon B, Gregorc V, et al. Mass spectrometry to classify non-small-cell lung cancer patients for clinical outcome after treatment with epidermal growth factor receptor tyrosine kinase inhibitors: a multicohort cross-institutional study. J Natl Cancer Inst 2007 Jun 6,99(11), 838-846.

(86) Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009 Jan,45(2), 228-247.

(87) Sorensen JB, Hansen HH, Hansen M, Dombernowsky P. Brain metastases in adenocarcinoma of the lung: frequency, risk groups, and prognosis. J Clin Oncol 1988 Sep,6(9), 1474-1480.

(88) Omuro AM, Kris MG, Miller VA, et al. High incidence of disease recurrence in the brain and leptomeninges in patients with nonsmall cell lung carcinoma after response to gefitinib. Cancer 2005 Jun 1,103(11), 2344-2348.

(89) Gleissner B, Chamberlain MC. Neoplastic meningitis. Lancet Neurol 2006 May,5(5), 443-452. (90) Ceresoli GL, Reni M, Chiesa G, et al. Brain metastases in locally advanced nonsmall cell lung

(21)

Referenties

GERELATEERDE DOCUMENTEN

Management of acquired resistance to epidermal growth factor receptor kinase inhibitors in patients with advanced non‐small cell lung cancer. Acquired resistance to tkis in

Doel van het onderzoek was het onderzoeken van de samenhang tussen depressie, angst en het zelfvertrouwen van vrouwen ten aanzien van de moederrol en in hoeverre inkomen, leeftijd

Acquired Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer..

Acquired Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer..

Acquired Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer..

Treatment of epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer (NSCLC) patients with tyrosine-kinase inhibitors (TKIs) results in high response rates

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are effective for leptomeningeal metastasis from non-small cell lung cancer patients with sensitive

Acquired Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer..